1
|
Tuohudaali W, Ji TF, Ding WT, Li CY, Bianba JS, Ci R, Zhao J. Urolithin B inhibits LPS-induced macrophage M1 polarization via miR155-5p mediated MAPK/NF-кB pathway. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-10. [PMID: 39671344 DOI: 10.1080/10286020.2024.2435984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
This study investigated inhibiting mechanisms of Urolithin B (Uro B) on macrophage M1 polarization. Uro B (50 μM) could inhibit the PGE2, COX-2, NO, iNOS, TNF-α, IL-1β and IL-6 levels compared with model group (P < 0.05) as well as the CD86 and F4/80 expression. The miR155-5p overexpression could increase the p38 MAPK, JNK, ERK mRNA activities (P < 0.05), Uro B (50 μM) could reverse changes in these indicators (P < 0.05). Moreover, Uro B (50 μM) could inhibit the TLR4, Src, IκBα, NF-κBp65 and their phosphorylated protein expression (P < 0.05). Therefore, Uro B may inhibit macrophage M1 polarization via miR155-5p mediated MAPK/NF-кB pathway.
Collapse
Affiliation(s)
| | - Teng-Fei Ji
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wan-Ting Ding
- School of Pharmacy, Xinjiang Medical University, Urumqi 830046, China
| | - Chen-Yang Li
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Urumqi 830004, China
| | | | - Ren Ci
- Hospital of Tibetan Traditional Medicine, Lhasa 850002, China
| | - Jun Zhao
- School of Pharmacy, Xinjiang Medical University, Urumqi 830046, China
- Key Laboratory for Uighur Medicine, Institute of Materia Medica of Xinjiang, Urumqi 830004, China
| |
Collapse
|
2
|
Massaga C, Paul L, Kwiyukwa LP, Vianney JM, Chacha M, Raymond J. Computational analysis of Urolithin A as a potential compound for anti-inflammatory, antioxidant, and neurodegenerative pathways. Free Radic Biol Med 2024; 227:508-520. [PMID: 39643139 DOI: 10.1016/j.freeradbiomed.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/24/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Urolithin A, an active precursor derived from the metabolism of ellagitanins in rats and humans, is known for its potential health benefits, including stimulating mitophagy and promoting muscular skeletal function. While experimental studies have demonstrated Urolithin A's potential to enhance cellular health, the detailed molecular interactions through which Urolithin A exerts its effects are not fully elucidated. In this study, we investigated the anti-inflammatory, antioxidation and neuroprotective abilities of Urolithin A in selected targets using molecular docking and molecular dynamics simulation methods. Molecular docking studies revealed the strong affinity for receptors involved in inflammation activities, including human p38 MAP kinase (4DLI) with -10.1 kcal/mol interacting with SER252, LYS249, and ASP294 residues. The binding energy in the 5KIR target was -8.6 kcal/mol, interacting with GLN203 through hydrogen bond, and lastly, 1A9U with an affinity of -6.8 with no hydrogen bond formed with Urolithin A and interacts with van der Waals interactions. In oxidant targets, the influence of Urolithin was observed in 1OG5 with -7.9 kcal/mol interacting with GLN185, PHE447. For the 1M17 target, the binding affinity was -7.7 kcal/mol interacting with THR95 residue and 1ZXM target at -7.4 kcal/mol interacting with TYR36, TYR216, and LEU234 residues. The neuroprotective ability of urolithin A was observed in selected targets for acetylcholinesterase; the binding energy was -9.7 kcal/mol interacting with van der Waals and π interactions; for the 1GQR target, the binding energy was -9.9 kcal/mol interacting with van der Waals and π interactions and for β-amylase (1iyt) the binding energy was -5.5 forming hydrogen bond with SER8, GLN15 residues. Molecular Dynamics simulations at 100 ns of Urolithin A compared with reference 4DLI. The Urolithin A-4DLI complex exhibited greater stability than the reference receptor, as confirmed by RMSD, RMSF, Radius of Gyration, Hydrogen bond, and SASA analyses.
Collapse
Affiliation(s)
- Caroline Massaga
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| | - Lucas Paul
- Department of Chemistry, Dar es Salaam University College of Education, P.O. Box 2329, Dar es Salaam, Tanzania.
| | - Lucas P Kwiyukwa
- Chemistry Department, College of Natural and Applied Sciences, University of Dar es Salaam, P.O. Box 35061, Dar es Salaam, Tanzania.
| | - John-Mary Vianney
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| | - Musa Chacha
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| | - Jofrey Raymond
- School of Life Science and Bioengineering, The Nelson Mandela African Institution of Science and Technology, P.O. Box 447, Arusha, Tanzania.
| |
Collapse
|
3
|
Ma C, Guo Q, Chen Y, Huang X, Hou L, Li D, Chen X, Chen F, Ma W. Pomegranate Juice and Its Bioactive Compounds: Promising Therapeutic Agents for Iron Deficiency Anemia. FOOD REVIEWS INTERNATIONAL 2024:1-23. [DOI: 10.1080/87559129.2024.2397514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Cuiping Ma
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Qing Guo
- Department of Hematology, Inner Mongolia International Mongolian Hospital, Inner Mongolia, China
| | - Yuhan Chen
- Department of Oncology and Hematology, Shenzhen Luohu District Hospital of Traditional Chinese Medicine, Guangdong, China
| | - Xiaohua Huang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Li Hou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Dongyun Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fei Chen
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Ma
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Lewko B, Wodzińska M, Daca A, Płoska A, Obremska K, Kalinowski L. Urolithin A Ameliorates the TGF Beta-Dependent Impairment of Podocytes Exposed to High Glucose. J Pers Med 2024; 14:914. [PMID: 39338168 PMCID: PMC11433157 DOI: 10.3390/jpm14090914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/30/2024] Open
Abstract
Increased activity of transforming growth factor-beta (TGF-β) is a key factor mediating kidney impairment in diabetes. Glomerular podocytes, the crucial component of the renal filter, are a direct target of TGF-β action, resulting in irreversible cell loss and progression of chronic kidney disease (CKD). Urolithin A (UA) is a member of the family of polyphenol metabolites produced by gut microbiota from ellagitannins and ellagic acid-rich foods. The broad spectrum of biological activities of UA makes it a promising candidate for the treatment of podocyte disorders. In this in vitro study, we investigated whether UA influences the changes exerted in podocytes by TGF-β and high glucose. Following a 7-day incubation in normal (NG, 5.5 mM) or high (HG, 25 mM) glucose, the cells were treated with UA and/or TGF-β1 for 24 h. HG and TGF-β1, each independent and in concert reduced expression of nephrin, increased podocyte motility, and up-regulated expression of b3 integrin and fibronectin. These typical-for-epithelial-to-mesenchymal transition (EMT) effects were inhibited by UA in both HG and NG conditions. UA also reduced the typically elevated HG expression of TGF-β receptors and activation of the TGF-β signal transducer Smad2. Our results indicate that in podocytes cultured in conditions mimicking the diabetic milieu, UA inhibits and reverses changes underlying podocytopenia in diabetic kidneys. Hence, UA should be considered as a potential therapeutic agent in podocytopathies.
Collapse
Affiliation(s)
- Barbara Lewko
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | | | - Agnieszka Daca
- Department of Physiopathology, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | | | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
- BioTechMed Center, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-223 Gdansk, Poland
| |
Collapse
|
5
|
Komatsu W, Kishi H, Uchiyama K, Ohhira S, Kobashi G. Urolithin A suppresses NLRP3 inflammasome activation by inhibiting the generation of reactive oxygen species and prevents monosodium urate crystal-induced peritonitis. Biosci Biotechnol Biochem 2024; 88:966-978. [PMID: 38772744 DOI: 10.1093/bbb/zbae068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/09/2024] [Indexed: 05/23/2024]
Abstract
The NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome triggers the maturation of interleukin-1β (IL-1β) and is implicated in the pathogenesis of various inflammatory diseases. Urolithin A, a gut microbial metabolite of ellagic acid, reportedly exerts antiinflammatory effects in vitro and in vivo. However, whether urolithin A suppresses NLRP3 inflammasome activation is unclear. In this study, urolithin A inhibited the cleavage of NLRP3 inflammasome agonist-induced caspase-1, maturation of IL-1β, and activation of pyroptosis in lipopolysaccharide-primed mouse bone marrow-derived macrophages. Urolithin A reduced generation of intracellular and mitochondrial reactive oxygen species (ROS) and restricted the interaction between thioredoxin-interacting protein and NLRP3, which attenuated NLRP3 inflammasome activation. Urolithin A administration prevented monosodium urate-induced peritonitis in mice. Collectively, these findings indicate that urolithin A suppresses NLRP3 inflammasome activation, at least partially, by repressing the generation of intracellular and mitochondrial ROS.
Collapse
Affiliation(s)
- Wataru Komatsu
- Department of Public Health, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Hisashi Kishi
- Department of Public Health, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Koji Uchiyama
- Department of Public Health, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Shuji Ohhira
- Department of Public Health, Dokkyo Medical University School of Medicine, Tochigi, Japan
| | - Gen Kobashi
- Department of Public Health, Dokkyo Medical University School of Medicine, Tochigi, Japan
| |
Collapse
|
6
|
Isla KKY, Tanae MM, de Lima-Landman MTR, de Magalhães PM, Lapa AJ, Souccar C. Vasorelaxant effects of ellagitannins isolated from Cuphea carthagenensis. PLANTA MEDICA 2024; 90:276-285. [PMID: 38272038 DOI: 10.1055/a-2240-7372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Cuphea carthagenensis (Jacq.) J. F. Macbr. is a popular plant in Brazilian folk medicine owing to its hypotensive and central nervous system depressant effects. This study aimed to validate the hypotensive effect of the plant's aqueous extract (AE) in rats and examine the vascular actions of three hydrolyzable tannins, oenothein B, woodfordin C, and eucalbanin B, isolated from AE. Systolic blood pressure in unanesthetized rats was determined using the non-invasive tail-cuff method. Oral treatment of normotensive rats with 0.5 and 1.0 g/kg/day AE induced a dose-related hypotensive effect after 1 week. In rat aortic rings pre-contracted with noradrenaline, all ellagitannins (20 - 180 µM) induced a concentration-related vasorelaxation. This effect was blocked by either removing the endothelium or pre-incubating with NG-nitro-l-arginine methyl ester (10 µM), an inhibitor of nitric oxide (NO) synthase. In KCl-depolarized rat portal vein preparations, the investigated compounds did not affect significantly the maximal contractile responses and pD2 values of the concentration-response curves to CaCl2. Our results demonstrated the hypotensive effect of C. carthagenensis AE in unanesthetized rats. All isolated ellagitannins induced vasorelaxation in vitro via activating NO synthesis/NO release from endothelial cells, without altering the Ca2+ influx in vascular smooth muscle preparations. Considering the low oral bioavailability of ellagitannins, the determined in vitro actions of these compounds are unlikely to account for the hypotensive effect of AE in vivo. It remains to be determined the role of the bioactive ellagitannin-derived metabolites in the hypotensive effect observed after oral treatment of unanesthetized rats with the plant extract.
Collapse
Affiliation(s)
- Kaori Katiuska Yamaguchi Isla
- Department of Pharmacology, Section of Natural Products, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Mirtes Midori Tanae
- Department of Pharmacology, Section of Natural Products, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Pedro Melillo de Magalhães
- Multidisciplinary Center for Chemical, Biological and Agricultural Research, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Antônio José Lapa
- Department of Pharmacology, Section of Natural Products, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Caden Souccar
- Department of Pharmacology, Section of Natural Products, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Ma L, Han T, Zhan YA. Mechanism and role of mitophagy in the development of severe infection. Cell Death Discov 2024; 10:88. [PMID: 38374038 PMCID: PMC10876966 DOI: 10.1038/s41420-024-01844-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
Mitochondria produce adenosine triphosphate and potentially contribute to proinflammatory responses and cell death. Mitophagy, as a conservative phenomenon, scavenges waste mitochondria and their components in the cell. Recent studies suggest that severe infections develop alongside mitochondrial dysfunction and mitophagy abnormalities. Restoring mitophagy protects against excessive inflammation and multiple organ failure in sepsis. Here, we review the normal mitophagy process, its interaction with invading microorganisms and the immune system, and summarize the mechanism of mitophagy dysfunction during severe infection. We highlight critical role of normal mitophagy in preventing severe infection.
Collapse
Affiliation(s)
- Lixiu Ma
- Department of Respiratory and Critical Care Medicine, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Tianyu Han
- Jiangxi Institute of Respiratory Disease, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yi-An Zhan
- Department of Respiratory and Critical Care Medicine, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
8
|
Jamialahmadi T, Hasanpour M, Vakilian F, Penson PE, Iranshahy M, Sahebkar A. Evaluation of Urolithin A Efficacy in Heart Failure Patients with Reduced Ejection Fraction: A Randomized, Double-blind, Crossover, Placebo-controlled Clinical Trial. Rev Recent Clin Trials 2024; 19:221-228. [PMID: 38415449 DOI: 10.2174/0115748871279354240209101604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Mitochondrial dysfunction and impaired mitophagy are integral to myocyte loss and the progression of heart failure. Urolithin A (UA), a microbiota-produced metabolite of ellagitannins and ellagic acid, is a known stimulator of mitophagy and mitochondrial biogenesis that has shown cardioprotective effects in experimental models. METHODS A randomized, double-blind, placebo-controlled 2×2 crossover trial was conducted on 10 patients with HF with reduced ejection fraction (HFrEF). The trial design involved two 4- week intervention periods of UA (500 mg BID) and placebo, separated by a 2-week washout phase. The patients underwent two-dimensional echocardiogram examination as well as blood sampling at the beginning and end of each period. RESULTS All patients completed the study. The results failed to reveal any significant effect of UA supplementation on echocardiographic measures (LVEF, LVEDD, LVESV, and TAPSE). Plasma concentrations of pro-BNP, glucose, and CRP (p >0.05) were also not altered. Serum HDL-C levels were increased with UA compared with placebo (+6.46 ± 2.33 mg/dL, p =0.026), whereas other lipid indices (LDL-C, triglycerides, total cholesterol, and VLDL-C) remained unchanged (p >0.05). CONCLUSION The results of the present study do not support any positive effect of UA supplementation in improving echocardiographic and biochemical indices of HFrEF. Further studies with higher doses of UA and longer supplementation duration are encouraged to be conducted.
Collapse
Affiliation(s)
- Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maede Hasanpour
- Department of Pharmacognosy and Medicinal Plants Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farveh Vakilian
- Atherosclerosis Prevention Research Centre, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Peter E Penson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Milad Iranshahy
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Nagabooshanam S, Kumar A, Ramamoorthy S, Saravanan N, Sundaramurthy A. Rapid and sensitive electrochemical detection of oxidized form of glutathione in whole blood samples using Bi-metallic nanocomposites. CHEMOSPHERE 2024; 346:140517. [PMID: 37879374 DOI: 10.1016/j.chemosphere.2023.140517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/07/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023]
Abstract
We report a facile one-pot synthesis of bimetallic nickel-gold (Ni-Au) nanocomposite for ultra-sensitive and selective electrochemical detection of oxidized glutathione (GSSG) by electrochemical deposition on fluorine doped tin oxide (FTO) substrate. The electrodeposition of Ni-Au nanocomposite on FTO was confirmed by various characterization techniques such as field emission scanning electron microscopy (FE-SEM), X-ray diffractometer (XRD) and Fourier transform infra-red (FTIR) spectroscopy. The cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS) was utilized for the electrochemical characterization of glutathione reductase (GR)/Ni-Au/FTO working electrode at each stage of modification. The GR enzyme immobilized on the Ni-Au/FTO working electrode via glutaraldehyde cross-linking exhibited excellent selectivity against GSSG in the presence of nicotinamide adenine dinucleotide phosphate (NADPH). The immobilized GR enzyme breaks down the GSSG to reduced glutathione (GSH) and converting NADPH to NADP+ whereby generating an electron for the electrochemical sensing of GSSG. The synergistic behavior of bimetals and good electro-catalytic property of the fabricated sensor provided a broad linear detection range from 1 fM to 1 μM with a limit of detection (LOD) of 6.8 fM, limit of quantification (LOQ) of 20.41 fM and sensitivity of 0.024 mA/μM/cm2. The interference with other molecules such as dopamine, glycine, ascorbic acid, uric acid and glucose was found to be negligible due to the better selectivity of GR enzyme towards GSSG. The shelf-life and response time of the fabricated electrode was found to be 30 days and 32 s, respectively. The real sample analysis of GSSG in whole blood samples showed average recovery percentage from 95 to 101% which matched well with the standard calibration plot of the fabricated sensor with relative standard deviation (RSD) below 10%.
Collapse
Affiliation(s)
- Shalini Nagabooshanam
- Biomaterials Research Laboratory (BMRL), Department of Chemical Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, Tamil Nadu, India; Department of Mechanical Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tenpaku-cho, Toyohashi, Aichi, 441-8580, Japan
| | - Akash Kumar
- Department of Physics and Nanotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, Tamil Nadu, India
| | - Sharmiladevi Ramamoorthy
- Department of Physics and Nanotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, Tamil Nadu, India
| | - Nishakavya Saravanan
- Department of Physics and Nanotechnology, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, Tamil Nadu, India
| | - Anandhakumar Sundaramurthy
- Biomaterials Research Laboratory (BMRL), Department of Chemical Engineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, 603203, Chengalpattu, Tamil Nadu, India.
| |
Collapse
|
10
|
Xiong R, Li B, Yu H, Fan T, Yu H, Yang Y, Wang JZ, Pi G, Yang X. Urolithin A Inhibits Anterior Basolateral Amygdala to Ventral Hippocampal CA1 Circuit to Ameliorate Amyloid-β-Impaired Social Ability. J Alzheimers Dis 2024; 99:1303-1316. [PMID: 38759018 DOI: 10.3233/jad-240298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
Background Anxiety and social withdrawal are highly prevalent among patients with Alzheimer's disease (AD). However, the neural circuit mechanisms underlying these symptoms remain elusive, and there is a need for effective prevention strategies. Objective This study aims to elucidate the neural circuitry mechanisms underlying social anxiety in AD. Methods We utilized 5xFAD mice and conducted a series of experiments including optogenetic manipulation, Tandem Mass Tag-labeled proteome analysis, behavioral assessments, and immunofluorescence staining. Results In 5xFAD mice, we observed significant amyloid-β (Aβ) accumulation in the anterior part of basolateral amygdala (aBLA). Behaviorally, 6-month-old 5xFAD mice displayed excessive social avoidance during social interaction. Concurrently, the pathway from aBLA to ventral hippocampal CA1 (vCA1) was significantly activated and exhibited a disorganized firing patterns during social interaction. By optogenetically inhibiting the aBLA-vCA1 pathway, we effectively improved the social ability of 5xFAD mice. In the presence of Aβ accumulation, we identified distinct changes in the protein network within the aBLA. Following one month of administration of Urolithin A (UA), we observed significant restoration of the abnormal protein network within the aBLA. UA treatment also attenuated the disorganized firings of the aBLA-vCA1 pathway, leading to an improvement in social ability. Conclusions The aBLA-vCA1 circuit is a vulnerable pathway in response to Aβ accumulation during the progression of AD and plays a crucial role in Aβ-induced social anxiety. Targeting the aBLA-vCA1 circuit and UA administration are both effective strategies for improving the Aβ-impaired social ability.
Collapse
Affiliation(s)
- Rui Xiong
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Binrui Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haitao Yu
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Fundamental Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Tianceng Fan
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiling Yu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guilin Pi
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xifei Yang
- Shenzhen Key Laboratory of Modern Toxicology, Shenzhen Medical Key Discipline of Health Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| |
Collapse
|
11
|
Revankar AG, Bagewadi ZK, Shaikh IA, Mannasaheb BA, Ghoneim MM, Khan AA, Asdaq SMB. In-vitro and computational analysis of Urolithin-A for anti-inflammatory activity on Cyclooxygenase 2 (COX-2). Saudi J Biol Sci 2023; 30:103804. [PMID: 37727526 PMCID: PMC10505678 DOI: 10.1016/j.sjbs.2023.103804] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
Cyclooxygenase 2 (COX-2) participates in the inflammation process by converting arachidonic acid into prostaglandin G2 which increases inflammation, pain and fever. COX-2 has an active site and a heme pocket and blocking these sites stops the inflammation. Urolithin A is metabolite of ellagitannin produced from humans and animals gut microbes. In the current study, Urolithin A showed good pharmacokinetic properties. Molecular docking of the complex of Urolithin A and COX-2 revealed the ligand affinity of -7.97 kcal/mol with the ligand binding sites at TYR355, PHE518, ILE517 and GLN192 with the 4-H bonds at a distance of 2.8 Å, 2.3 Å, 2.5 Å and 1.9 Å. The RMSD plot for Urolithin A and COX-2 complex was observed to be constant throughout the duration of dynamics. A total of 3 pair of hydrogen bonds was largely observed on average of 3 simulation positions for dynamics duration of 500 ns. The MMPBSA analysis showed that active site amino acids had a binding energy of -22.0368 kJ/mol indicating that throughout the simulation the protein of target was bounded by Urolithin A. In-silico results were validated by biological assays. Urolithin A strongly revealed to exhibit anti-inflammatory effect on COX-2 with an IC50 value of 44.04 µg/mL. The anti-inflammatory capability was also depicted through reduction of protein denaturation that showed 37.6 ± 0.1 % and 43.2 ± 0.07 % reduction of protein denaturation for BSA and egg albumin respectively at 500 µg/mL. The present study, suggests Urolithin A to be an effective anti-inflammatory compound for therapeutic use.
Collapse
Affiliation(s)
- Archana G. Revankar
- Department of Biotechnology, KLE Technological University, Hubballi, Karnataka 580031, India
| | - Zabin K. Bagewadi
- Department of Biotechnology, KLE Technological University, Hubballi, Karnataka 580031, India
| | - Ibrahim Ahmed Shaikh
- Department of Pharmacology, College of Pharmacy, Najran University, Najran 66462, Saudi Arabia
| | | | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah 13713, Saudi Arabia
| | - Aejaz Abdullatif Khan
- Department of General Science, Ibn Sina National College for Medical Studies, Jeddah 21418, Saudi Arabia
| | | |
Collapse
|
12
|
Orabi MAA, Hasan AH, AbouZid SF, El Amir D, Hetta MH, Awadh AAA, Alqahtani OS, Hatano T, El-Shanawany MA. Nutritional, Antioxidant, Antimicrobial, and Anticholinesterase Properties of Phyllanthus emblica: A Study Supported by Spectroscopic and Computational Investigations. Metabolites 2023; 13:1013. [PMID: 37755293 PMCID: PMC10537340 DOI: 10.3390/metabo13091013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
Dietary fruits and vegetables play a vital role as food and drugs and are the main sources of antioxidant defences against degenerative diseases, such as brain dysfunctions, cardiovascular diseases, immune system deteriorations, and cancers, brought on by oxidative damage. Phyllanthus emblica is a significant herbal remedy used in conventional medicine to recover lost strength and power. In this research, the potential value of Phyllanthus emblica as a food and drug is researched. The total phenolic, total flavonoid, and total tannin contents as well as the nutritional value, vitamin C, vitamin E, and mineral contents of different organs of P. emblica were evaluated. The antioxidant and antimicrobial activities of extracts and fractions of different organs of P. emblica were determined. A total of eleven flavonoids, simple phenolic, tannin-related phenolic, and tannin molecules were isolated from a hydroalcoholic extract of the leaves and fruits. The structures were identified by spectroscopic data and comparison with the literature values as gallic acid (1), naringenin 7-O-(6″-O-galloyl)-β-D-glucopyranoside (2), 3,3'-di-O-methyl ellagic acid-4'-O-β-d-glucopyranoside (3), 1-O-galloyl glycerol (4), 1,6-di-O-galloyl-β-d-glucopyranoside (5), flavogallonic acid bislactone (6), corilagin (7), ethyl gallate (8), urolithin M5 (9), (E)-p-coumaroyl-1-O-β-d-glucopyranoside (10), and 1,2,4,6-tetra-O-galloyl-β-d-glucopyranoside (11). Among them, compounds 3 and 10 are first isolated from the plant. Molecular docking was performed to investigate the comparative interactions between positive controls (galantamine and donepezil) and selected compounds utilizing acetylcholinesterase (4EY7) as a target receptor. Results exhibited the potency of these compounds against the target receptor. In summary, P. emblica has a wealth of minerals, vitamins C and E, and polyphenolic phytochemicals that may work together to treat infectious disease, prevent and/or treat oxidative-damage-related illnesses including Alzheimer's disease.
Collapse
Affiliation(s)
- Mohamed A. A. Orabi
- Department of Pharmacognosy, College of Pharmacy, Najran University, Najran 66454, Saudi Arabia;
| | - Aso Hameed Hasan
- Department of Chemistry, College of Science, University of Garmian, Kalar 46021, Kurdistan Region, Iraq;
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Johor Bahru 81310, Johor, Malaysia
| | - Sameh F. AbouZid
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt;
| | - Dalia El Amir
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt;
| | - Mona H. Hetta
- Department of Pharmacognosy, Faculty of Pharmacy, Fayoum University, Fayoum 63514, Egypt;
| | - Ahmed Abdullah Al Awadh
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran 66454, Saudi Arabia;
| | - Omaish Salman Alqahtani
- Department of Pharmacognosy, College of Pharmacy, Najran University, Najran 66454, Saudi Arabia;
| | - Tsutomu Hatano
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Tsushima, Okayama 700-8530, Japan;
| | - Mohamed A. El-Shanawany
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City 11829, Egypt;
| |
Collapse
|
13
|
An L, Lu Q, Wang K, Wang Y. Urolithins: A Prospective Alternative against Brain Aging. Nutrients 2023; 15:3884. [PMID: 37764668 PMCID: PMC10534540 DOI: 10.3390/nu15183884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
The impact of host-microbiome interactions on cognitive health and disease has received increasing attention. Microbial-derived metabolites produced in the gut are one of crucial mechanisms of the gut-brain axis interaction, showing attractive perspectives. Urolithins (Uros) are gut microbial-derived metabolites of ellagitannins and ellagic acid, whose biotransformation varies considerably between individuals and decreases greatly with age. Recently, accumulating evidence has suggested that Uros may have specific advantages in preventing brain aging including favorable blood-brain barrier permeability, selective brain distribution, and increasingly supporting data from preclinical and clinical studies. However, the usability of Uros in diagnosis, prevention, and treatment of neurodegenerative diseases remains elusive. In this review, we aim to present the comprehensive achievements of Uros in age-related brain dysfunctions and neurodegenerative diseases and discuss their prospects and knowledge gaps as functional food, drugs, or biomarkers against brain aging.
Collapse
Affiliation(s)
- Lei An
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (L.A.); (Q.L.); (K.W.)
| | - Qiu Lu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (L.A.); (Q.L.); (K.W.)
| | - Ke Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (L.A.); (Q.L.); (K.W.)
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
- Rizhao Huawei Institute of Comprehensive Health Industries, Shandong Keepfit Biotech. Co., Ltd., Rizhao 276800, China
| | - Yousheng Wang
- College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| |
Collapse
|
14
|
Boeing T, Reis Lívero FAD, de Souza P, de Almeida DAT, Donadel G, Lourenço ELB, Gasparotto Junior A. Natural Products as Modulators of Mitochondrial Dysfunctions Associated with Cardiovascular Diseases: Advances and Opportunities. J Med Food 2023; 26:279-298. [PMID: 37186894 DOI: 10.1089/jmf.2022.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
The mitochondria have an important role in modulating cell cycle progression, cell survival, and apoptosis. In the adult heart, the cardiac mitochondria have a unique spatial arrangement and occupy nearly one-third the volume of a cardiomyocyte, being highly efficient for converting the products of glucose or fatty acid metabolism into adenosine triphosphate (ATP). In cardiomyocytes, the decline of mitochondrial function reduces ATP generation and increases the production of reactive oxygen species, which generates impaired heart function. This is because mitochondria play a key role in maintaining cytosolic calcium concentration and modulation of muscle contraction, as ATP is required to dissociate actin from myosin. Beyond that, mitochondria have a significant role in cardiomyocyte apoptosis because it is evident that patients who have cardiovascular diseases (CVDs) have increased mitochondrial DNA damage to the heart and aorta. Many studies have shown that natural products have mitochondria-modulating effects in cardiac diseases, determining them as potential candidates for new medicines. This review outlines the leading plant secondary metabolites and natural compounds derived from microorganisms as modulators of mitochondrial dysfunctions associated with CVDs.
Collapse
Affiliation(s)
- Thaise Boeing
- Graduate Program in Pharmaceutical Sciences, Chemical-Pharmaceutical Research Nucleus, University of Vale do Itajaí, Itajaí, Brazil
| | - Francislaine Aparecida Dos Reis Lívero
- Laboratory of Pre-Clinical Research of Natural Products, Postgraduate Program in Animal Science with Emphasis on Bioactive Products, Paranaense University, Umuarama, Brazil
| | - Priscila de Souza
- Graduate Program in Pharmaceutical Sciences, Chemical-Pharmaceutical Research Nucleus, University of Vale do Itajaí, Itajaí, Brazil
| | - Danielle Ayr Tavares de Almeida
- Laboratory of Cardiovascular Pharmacology (LaFaC), Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Brazil
| | - Guilherme Donadel
- Laboratory of Pre-Clinical Research of Natural Products, Postgraduate Program in Animal Science with Emphasis on Bioactive Products, Paranaense University, Umuarama, Brazil
| | - Emerson Luiz Botelho Lourenço
- Laboratory of Pre-Clinical Research of Natural Products, Postgraduate Program in Animal Science with Emphasis on Bioactive Products, Paranaense University, Umuarama, Brazil
| | - Arquimedes Gasparotto Junior
- Laboratory of Cardiovascular Pharmacology (LaFaC), Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Brazil
| |
Collapse
|
15
|
Augustynowicz D, Lemieszek MK, Strawa JW, Wiater A, Tomczyk M. Phytochemical Profiling of Extracts from Rare Potentilla Species and Evaluation of Their Anticancer Potential. Int J Mol Sci 2023; 24:ijms24054836. [PMID: 36902263 PMCID: PMC10002591 DOI: 10.3390/ijms24054836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Despite the common use of Potentilla L. species (Rosaceae) as herbal medicines, a number of species still remain unexplored. Thus, the present study is a continuation of a study evaluating the phytochemical and biological profiles of aqueous acetone extracts from selected Potentilla species. Altogether, 10 aqueous acetone extracts were obtained from the aerial parts of P. aurea (PAU7), P. erecta (PER7), P. hyparctica (PHY7), P. megalantha (PME7), P. nepalensis (PNE7), P. pensylvanica (PPE7), P. pulcherrima (PPU7), P. rigoi (PRI7), and P. thuringiaca (PTH7), leaves of P. fruticosa (PFR7), as well as from the underground parts of P. alba (PAL7r) and P. erecta (PER7r). The phytochemical evaluation consisted of selected colourimetric methods, including total phenolic (TPC), tannin (TTC), proanthocyanidin (TPrC), phenolic acid (TPAC), and flavonoid (TFC) contents, as well as determination of the qualitative secondary metabolite composition by the employment of LC-HRMS (liquid chromatography-high-resolution mass spectrometry) analysis. The biological assessment included an evaluation of the cytotoxicity and antiproliferative properties of the extracts against human colon epithelial cell line CCD841 CoN and human colon adenocarcinoma cell line LS180. The highest TPC, TTC, and TPAC were found in PER7r (326.28 and 269.79 mg gallic acid equivalents (GAE)/g extract and 263.54 mg caffeic acid equivalents (CAE)/g extract, respectively). The highest TPrC was found in PAL7r (72.63 mg catechin equivalents (CE)/g extract), and the highest TFC was found in PHY7 (113.29 mg rutin equivalents (RE)/g extract). The LC-HRMS analysis showed the presence of a total of 198 compounds, including agrimoniin, pedunculagin, astragalin, ellagic acid, and tiliroside. An examination of the anticancer properties revealed the highest decrease in colon cancer cell viability in response to PAL7r (IC50 = 82 µg/mL), while the strongest antiproliferative effect was observed in LS180 treated with PFR7 (IC50 = 50 µg/mL) and PAL7r (IC50 = 52 µg/mL). An LDH (lactate dehydrogenase) assay revealed that most of the extracts were not cytotoxic against colon epithelial cells. At the same time, the tested extracts for the whole range of concentrations damaged the membranes of colon cancer cells. The highest cytotoxicity was observed for PAL7r, which in concentrations from 25 to 250 µg/mL increased LDH levels by 145.7% and 479.0%, respectively. The previously and currently obtained results indicated that some aqueous acetone extracts from Potentilla species have anticancer potential and thus encourage further studies in order to develop a new efficient and safe therapeutic strategy for people who have been threatened by or suffered from colon cancer.
Collapse
Affiliation(s)
- Daniel Augustynowicz
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
| | - Marta Kinga Lemieszek
- Department of Medical Biology, Institute of Rural Health, ul. Jaczewskiego 2, 20-090 Lublin, Poland
| | - Jakub Władysław Strawa
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
| | - Adrian Wiater
- Department of Industrial and Environmental Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, ul. Akademicka 19, 20-033 Lublin, Poland
| | - Michał Tomczyk
- Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
- Correspondence: ; Tel.: +48-85-748-56-94
| |
Collapse
|
16
|
Daschner PJ, Ross S, Seifried H, Kumar A, Flores R. Nutrition and Microbiome Interactions in Human Cancer. J Acad Nutr Diet 2023; 123:504-514. [PMID: 36208721 DOI: 10.1016/j.jand.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/11/2022]
Abstract
Individual physiologic responses to changes in dietary patterns can vary widely to affect cancer risk, which is driven by multiple host-specific factors (eg, genetics, epigenetics, inflammatory and metabolic states, and the colonizing microbiome). Emerging evidence indicates that diet-induced microbiota alterations are key modulators of several host functions important to tumor etiology, progression, and response to cancer therapy. Thus, diet may potentially be used to target alterations of the microbiota as an effective means to improve outcomes across the cancer continuum (from cancer prevention to tumor development and progression, to effects on treatment and survivorship). This review will focus on recent examples of functional interactions between dietary components (nutrients and non-nutrients) and the gastrointestinal microbiome, which are 2 critical and malleable environmental variables in cancer risk that affect host immune, metabolic, and cell signaling functions and may provide insights for novel cancer therapeutic and preventive strategies.
Collapse
Affiliation(s)
- Phillip J Daschner
- Division of Cancer Biology, National Cancer Institute, Bethesda, Maryland.
| | - Sharon Ross
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Harold Seifried
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Amit Kumar
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Roberto Flores
- Office of Nutrition Research, Division of Program Coordination, Planning and Strategic Initiatives, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
17
|
Pantiora PD, Balaouras AI, Mina IK, Freris CI, Pappas AC, Danezis GP, Zoidis E, Georgiou CA. The Therapeutic Alliance between Pomegranate and Health Emphasizing on Anticancer Properties. Antioxidants (Basel) 2023; 12:187. [PMID: 36671048 PMCID: PMC9855163 DOI: 10.3390/antiox12010187] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Pomegranate is a fruit bearing-plant that is well known for its medicinal properties. Pomegranate is a good source of phenolic acids, tannins, and flavonoids. Pomegranate juice and by-products have attracted the scientific interest due to their potential health benefits. Currently, the medical community has showed great interest in exploiting pomegranate potential as a protective agent against several human diseases including cancer. This is demonstrated by the fact that there are more than 800 reports in the literature reporting pomegranate's anticancer properties. This review is an update on the research outcomes of pomegranate's potential against different types of human diseases, emphasizing on cancer. In addition, perspectives of potential applications of pomegranate, as a natural additive aiming to improve the quality of animal products, are discussed.
Collapse
Affiliation(s)
- Panagiota D. Pantiora
- Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | | | - Ioanna K. Mina
- Department of Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Christoforos I. Freris
- Department of Chemistry, National and Kapodistrian University of Athens, 15784 Athens, Greece
| | - Athanasios C. Pappas
- Laboratory of Nutritional Physiology and Feeding, Department of Animal Science, Agricultural University of Athens, 11855 Athens, Greece
| | - Georgios P. Danezis
- Chemistry Laboratory, Department of Food Science and Human Nutrition, Agricultural University of Athens, 11855 Athens, Greece
| | - Evangelos Zoidis
- Laboratory of Nutritional Physiology and Feeding, Department of Animal Science, Agricultural University of Athens, 11855 Athens, Greece
| | - Constantinos A. Georgiou
- Chemistry Laboratory, Department of Food Science and Human Nutrition, Agricultural University of Athens, 11855 Athens, Greece
| |
Collapse
|
18
|
Li J, Huang X, He L, Li C, Jing H, Lin J, Ma C, Li X. Effect of ellagic acid on body weight, nutrient digestibility, fecal microbiota, and urolithin A metabolism in Thoroughbred horses. J Anim Sci 2023; 101:skad232. [PMID: 37422771 PMCID: PMC10612130 DOI: 10.1093/jas/skad232] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 07/07/2023] [Indexed: 07/11/2023] Open
Abstract
This study aimed to investigate the effects of ellagic acid (EA) supplementation on body weight (BW), nutrient digestibility, fecal microbiota, blood biochemical indices, and urolithin A metabolism in 1-yr-old Thoroughbred horses. A group of 18 1-yr-old Thoroughbred horses, with an average weight of 339.00 ± 30.11 kg, were randomly allocated into three groups of six horses each (three males and three females). The control group (n = 6) received only the basal diet, whereas test groups I (n = 6) and II (n = 6) were fed the basal diet supplemented with 15 mg/kg BW/d and 30 mg/kg BW/d of EA, respectively, for 40-d. The results showed that test group I and II horses had a significant increase in total weight gain by 49.47% and 62.74%, respectively, compared to the control group. The digestibility of various components in the diets of the test group horses was improved, including dry matter, organic matter, gross energy, neutral detergent fiber, acid detergent fiber, and calcium. Additionally, the digestibility of crude protein and phosphorus (P) in test group II horses increased significantly by 10.96% and 33.56% (P < 0.05), respectively. Moreover, EA supplementation significantly increased the fecal abundance of Firmicutes, Bacteroidetes (P < 0.05), Fibrobacterota, p-251-o5, Desemzia incerta (P < 0.05), and Fibrobacter sp. (P < 0.05), while reducing the abundance of Proteobacteria, Pseudomonadaceae, Pseudomonas, and Cupriavidus pauculus (P < 0.05 or P < 0.01). Fecal samples from test group II showed 89.47%, 100%, and 86.15% increases in the concentrations of acetic acid, valeric acid, and total volatile fatty acids, respectively. In addition, the plasma levels of total protein, and globulin increased significantly in test groups I (7.88% and 11.35%, respectively) and II (13.44% and 16.07%, respectively) compared to those in the control group (P < 0.05). The concentration of urolithin A in fecal and urine samples was positively correlated with increasing doses of EA. These findings suggest that supplemental feeding of EA improved nutrient digestibility, blood biochemical indices, and fecal microbiota in 1-yr-old Thoroughbred horses, promoting growth and development.
Collapse
Affiliation(s)
- Jiahao Li
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| | - Xinxin Huang
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| | - Linjiao He
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| | - Chao Li
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| | - Hongxin Jing
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| | - Jianwei Lin
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| | - Chaoyu Ma
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| | - Xiaobin Li
- College of Animal Science, Xinjiang Key Laboratory of Herbivore Nutrition for Meat & Milk Production, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
- College of Animal Science, Xinjiang Key Laboratory of Horse Breeding and Exercise Physiology, Xinjiang Agricultural University, Urumqi, Xinjiang 830052, China
| |
Collapse
|
19
|
Looi D, Moorthy M, Chaiyakunapruk N, Devi Palanisamy U. Impact of ellagitannin-rich fruit consumption on blood pressure: A systematic review and meta-analysis of randomized controlled trials. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
20
|
Mi H, Liu S, Hai Y, Yang G, Lu J, He F, Zhao Y, Xia M, Hou X, Fang Y. Lactococcus garvieae FUA009, a Novel Intestinal Bacterium Capable of Producing the Bioactive Metabolite Urolithin A from Ellagic Acid. Foods 2022; 11:2621. [PMID: 36076807 PMCID: PMC9455165 DOI: 10.3390/foods11172621] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/18/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Dietary polyphenol ellagic acid has anti-cancer and anti-inflammatory activities, and these biological activities require the conversion of ellagic acid to urolithins by intestinal microbes. However, few gut microbes are capable of metabolizing ellagic acid to produce urolithins, limiting the beneficial effects of ellagic acid on health. Here, we describe an intestinal bacterium Lactococcus garvieae FUA009 isolated from the feces of a healthy volunteer. It was demonstrated via HPLC and UPLC-MS analysis that the end product of ellagic acid metabolism of FUA009 was urolithin A. In addition, we also examined the whole genome sequence of FUA009 and then assessed the safety and probiotic properties of FUA009 based on a complete genome and phenotype analysis. We indicated that FUA009 was safe, which was confirmed by FUA009 being sensitive to multiple antibiotics, having no hemolytic activity, and being free of aggressive putative virulence factors. Moreover, 19 stress-responsive protein genes and 8 adhesion-related genes were predicted in the FUA009 genome. Furthermore, we demonstrated that FUA009 was tolerant to acid and bile salt by determining the cell viability in a stress environment. In summary, Lactococcus garvieae FUA009, as a novel UA-producing bacterium, not only contributes to the study of the metabolic pathway of ellagic acid but is also expected to be a novel probiotic candidate.
Collapse
Affiliation(s)
- Haoyu Mi
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Shu Liu
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
- College of Food Science and Engineering, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yang Hai
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Guang Yang
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
- College of Food Science and Engineering, Jiangsu Ocean University, Lianyungang 222005, China
| | - Jing Lu
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
- College of Food Science and Engineering, Jiangsu Ocean University, Lianyungang 222005, China
| | - Fuxiang He
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Yaling Zhao
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Mengjie Xia
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
| | - Xiaoyue Hou
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
- College of Food Science and Engineering, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Marine Resources Development Research Institute, Jiangsu Ocean University, Lianyungang 222005, China
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yaowei Fang
- Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang 222005, China
- Jiangsu Marine Resources Development Research Institute, Jiangsu Ocean University, Lianyungang 222005, China
| |
Collapse
|
21
|
Boozari M, Hosseinzadeh H. Crocin molecular signaling pathways at a glance: A comprehensive review. Phytother Res 2022; 36:3859-3884. [PMID: 35989419 DOI: 10.1002/ptr.7583] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/16/2022] [Accepted: 07/07/2022] [Indexed: 11/09/2022]
Abstract
Crocin is a hydrophilic carotenoid that is synthesized in the flowers of the Crocus genus. Numerous in vitro and in vivo research projects have been published about the biological and pharmacological properties and toxicity of crocin. Crocin acts as a memory enhancer, anxiolytic, aphrodisiac, antidepressant, neuroprotective, and so on. Here, we introduce an updated and comprehensive review of crocin molecular mechanisms based on previously examined and mentioned in the literature. Different studies confirmed the significant effect of crocin to control pathological conditions, including oxidative stress, inflammation, metabolic disorders, neurodegenerative disorders, and cancer. The neuroprotective effect of crocin could be related to the activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT)/mammalian target of rapamycin (mTOR), Notch, and cyclic-AMP response element-binding protein signaling pathways. The crocin also protects the cardiovascular system through the inhibitory effect on toll-like receptors. The regulatory effect of crocin on PI3K/AKT/mTOR, AMP-activated protein kinase, mitogen-activated protein kinases (MAPK), and peroxisome proliferator-activated receptor pathways can play an effective role in the treatment of metabolic disorders. The crocin has anticancer activity through the PI3K/AKT/mTOR, MAPK, vascular endothelial growth factor, Wnt/β-catenin, and Janus kinases-signal transducer and activator of transcription suppression. Also, the nuclear factor-erythroid factor 2-related factor 2 and p53 signaling pathway activation may be effective in the anticancer effect of crocin. Finally, among signaling pathways regulated by crocin, the most important ones seem to be those related to the regulatory effect on the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Motahareh Boozari
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
22
|
Kotewicz M, Krauze-Baranowska M, Daca A, Płoska A, Godlewska S, Kalinowski L, Lewko B. Urolithins Modulate the Viability, Autophagy, Apoptosis, and Nephrin Turnover in Podocytes Exposed to High Glucose. Cells 2022; 11:cells11162471. [PMID: 36010548 PMCID: PMC9406555 DOI: 10.3390/cells11162471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/06/2022] [Indexed: 12/02/2022] Open
Abstract
Urolithins are bioactive compounds generated in human and animal intestines because of the bacterial metabolism of dietary ellagitannins (and their constituent, ellagic acid). Due to their multidirectional effects, including anti-inflammatory, antioxidant, anti-cancer, neuroprotective, and antiglycative properties, urolithins are potential novel therapeutic agents. In this study, while considering the future possibility of using urolithins to improve podocyte function in diabetes, we assessed the results of exposing mouse podocytes cultured in normal (NG, 5.5 mM) and high (HG, 25 mM) glucose concentrations to urolithin A (UA) and urolithin B (UB). Podocytes metabolized UA to form glucuronides in a time-dependent manner; however, in HG conditions, the metabolism was lower than in NG conditions. In HG milieu, UA improved podocyte viability more efficiently than UB and reduced the reactive oxygen species level. Both types of urolithins showed cytotoxic activity at high (100 µM) concentration. The UA upregulated total and surface nephrin expression, which was paralleled by enhanced nephrin internalization. Regulation of nephrin turnover was independent of ambient glucose concentration. We conclude that UA affects podocytes in different metabolic and functional aspects. With respect to its pro-survival effects in HG-induced toxicity, UA could be considered as a potent therapeutic candidate against diabetic podocytopathy.
Collapse
Affiliation(s)
- Milena Kotewicz
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | | | - Agnieszka Daca
- Department of Pathology and Experimental Rheumatology, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Sylwia Godlewska
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-233 Gdansk, Poland
| | - Barbara Lewko
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
- Correspondence:
| |
Collapse
|
23
|
Zhang C, Song Y, Chen L, Chen P, Yuan M, Meng Y, Wang Q, Zheng G, Qiu Z. Urolithin A Attenuates Hyperuricemic Nephropathy in Fructose-Fed Mice by Impairing STING-NLRP3 Axis-Mediated Inflammatory Response via Restoration of Parkin-Dependent Mitophagy. Front Pharmacol 2022; 13:907209. [PMID: 35784701 PMCID: PMC9240289 DOI: 10.3389/fphar.2022.907209] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/26/2022] [Indexed: 11/23/2022] Open
Abstract
Urolithin A (UroA) is one of the primary intestinal metabolites of ellagitannins, showing translational potential as a nutritional intervention in humans. Mounting evidence suggests that fructose consumption contributes to the progression of chronic kidney disease (CKD) that manifests in hyperuricemic nephropathy, renal inflammation, and tubulointerstitial injury. Here, we investigated the efficacy of UroA in alleviating fructose-induced hyperuricemic nephropathy in mice. Uric acid-exposed human kidney-2 (HK-2) cells were utilized for in vitro mechanism validation. Histopathological staining, immunoblotting, and transmission electron microscope were performed for the mechanistic investigations. Our results revealed that UroA ameliorated fructose-induced hyperuricemic nephropathy in mice. The histopathologic assessment showed that UroA attenuated tubular hypertrophy and dilation, glomerular basement membrane thickening, and collagen deposition in the kidney of fructose-fed mice. Mechanistically, UroA treatment impaired STING-NLRP3 activation, resulting in reduced production of proinflammatory cytokines IL-1β, IL-6, and TNF-α. Notably, UroA exhibited a scavenging effect against reactive oxygen species (ROS) and restored fructose-impaired PINK1/Parkin-mediated mitophagy in nephropathic mice. Furthermore, the inhibitory effect of UroA in STING-NLRP3 activation was impaired after Parkin gene silencing in HK-2 cells. Together, this study suggests that UroA alleviates fructose-induced hyperuricemic nephropathy by promoting Parkin-dependent mitophagy, thereby suppressing STING-NLRP3 axis-mediated inflammatory response. Thus, dietary supplementation with UroA or ellagitannins-rich foods may serve as a promising intervention to prevent CKD progression.
Collapse
Affiliation(s)
- Cong Zhang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yingying Song
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Liang Chen
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Peng Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ming Yuan
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Yan Meng
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Qi Wang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Guohua Zheng
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China
- *Correspondence: Zhenpeng Qiu, ; Guohua Zheng,
| | - Zhenpeng Qiu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan, China
- *Correspondence: Zhenpeng Qiu, ; Guohua Zheng,
| |
Collapse
|
24
|
Tao H, Tao Y, Yang C, Li W, Zhang W, Li X, Gu Y, Hong Y, Yang H, Liu Y, Yang X, Geng D. Gut Metabolite Urolithin A Inhibits Osteoclastogenesis and Senile Osteoporosis by Enhancing the Autophagy Capacity of Bone Marrow Macrophages. Front Pharmacol 2022; 13:875611. [PMID: 35645801 PMCID: PMC9135380 DOI: 10.3389/fphar.2022.875611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/29/2022] [Indexed: 12/16/2022] Open
Abstract
Senile osteoporosis (SOP) is a systemic bone disease that is significantly associated with age and eventually leads to deteriorated bone strength and increased fracture risk. Urolithin A (Uro-A) is a gut microbiome-derived compound that is mainly produced from pomegranates and some nuts. Uro-A has attracted great attention in recent years in view of its protective effects on aging-related diseases, including muscle dysfunction, kidney disease and knee injury. However, its protective influence and possible mechanisms in senile osteoporosis remain unclear. Our study describes the beneficial effect of Uro-A on bone marrow macrophages (BMMs). The in vitro results demonstrated that Uro-A inhibited receptor activator for nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis in BMMs in a concentration-dependent manner. Uro-A significantly reduced the expression of osteoclast-related genes and bone resorption. Mechanistically, we found that the autophagy ability of BMMs was significantly enhanced in the early stage of Uro-A treatment, accompanied by the activation of LC3 and Beclin 1. At the same time, this enhanced autophagy activity was maintained until the later stage after stimulation with RANKL. Furthermore, we found that the MARK signaling pathway was blocked by Uro-A treatment. In a mouse model of aging, Uro-A effectively inhibited bone loss in the proximal femur, spine and tibia of aging mice. These results indicated that Uro-A is a robust and effective treatment for preventing senile osteoporosis bone loss.
Collapse
Affiliation(s)
- Huaqiang Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yunxia Tao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chen Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueyan Li
- Anesthesiology Department, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Ye Gu
- Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, China
| | - Yujing Hong
- Department of Preventive Medicine, Nantong University, Nantong, China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Liu
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, China
| | - Xing Yang
- Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Orthopedics, Changshu Hospital Affiliated to Soochow University, First People's Hospital of Changshu City, Changshu, China
| |
Collapse
|
25
|
Liu JK. Antiaging agents: safe interventions to slow aging and healthy life span extension. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:18. [PMID: 35534591 PMCID: PMC9086005 DOI: 10.1007/s13659-022-00339-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/29/2022] [Indexed: 05/02/2023]
Abstract
Human longevity has increased dramatically during the past century. More than 20% of the 9 billion population of the world will exceed the age of 60 in 2050. Since the last three decades, some interventions and many preclinical studies have been found to show slowing aging and increasing the healthy lifespan of organisms from yeast, flies, rodents to nonhuman primates. The interventions are classified into two groups: lifestyle modifications and pharmacological/genetic manipulations. Some genetic pathways have been characterized to have a specific role in controlling aging and lifespan. Thus, all genes in the pathways are potential antiaging targets. Currently, many antiaging compounds target the calorie-restriction mimetic, autophagy induction, and putative enhancement of cell regeneration, epigenetic modulation of gene activity such as inhibition of histone deacetylases and DNA methyltransferases, are under development. It appears evident that the exploration of new targets for these antiaging agents based on biogerontological research provides an incredible opportunity for the healthcare and pharmaceutical industries. The present review focus on the properties of slow aging and healthy life span extension of natural products from various biological resources, endogenous substances, drugs, and synthetic compounds, as well as the mechanisms of targets for antiaging evaluation. These bioactive compounds that could benefit healthy aging and the potential role of life span extension are discussed.
Collapse
Affiliation(s)
- Ji-Kai Liu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, People's Republic of China.
| |
Collapse
|
26
|
Mao C, Luan H, Gao S, Sheng W. Urolithin A as a Potential Drug for the Treatment of Spinal Cord Injuries: A Mechanistic Study Using Network Pharmacology Approaches. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:9090113. [PMID: 35497925 PMCID: PMC9054438 DOI: 10.1155/2022/9090113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/08/2022] [Accepted: 03/21/2022] [Indexed: 02/08/2023]
Abstract
Objective This research was focused to examine the potential targets, action network, and mechanism of urolithin A (UA) in spinal cord injury (SCI) management exploiting the network pharmacology (NP). Methods We used the SwissTargetPrediction, PharmMapper, and TargetNet databases to obtain UA action targets. We searched the OMIM, GeneCards, CTD, and DrugBank databases to screen selected target genes for SCI treatment. The intersection of target genes between the UA and SCI databases was obtained by constructing Venn diagrams, which led to the identification of common druggable targets for the disease. The relationship network of the targets was built with Cytoscape 3.7.2, and the protein interaction network was analyzed with the STRING platform. The protein-protein interaction (PPI) network can be built on the STRING database. Gene Ontology (GO) function and KEGG pathway analyses of target intersections were completed with the DAVID 6.8 database. We constructed preliminary network targets for actions underlying UA-SCI interactions. Using the AutoDock software, we examined the molecular docking interactions between UA and its target proteins and further verified the mechanism of the action of UA. Results We obtained 318 UA drug targets and 1492 SCI disease targets. We identified a total of 118 common UA-SCI targets. Based on the PPI analysis, we identified MAPK1, SRC, AKT1, HRAS, MAPK8, HSP90AA1, MAPK14, JAK2, ESR1, and NF-κB1 as possible therapeutic targets. Enrichment analysis revealed that the PI3K-AKT, VEGF, and TNF signaling pathways could be critical for the NP analysis. Molecular docking indicated that UA had a strong affinity for docked proteins (binding energy range: -6.3 to -9.3 kcal mol-1). Conclusions We employed an NP approach to validate and predict the underlying mechanisms associated with UA therapy for SCI. An additional purpose of this study was to provide a theoretical basis for further experimental studies on UA's potential in SCI treatment.
Collapse
Affiliation(s)
- Chao Mao
- Department of Spine Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - HaoPeng Luan
- Department of Spine Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - ShuTao Gao
- Department of Spine Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| | - WeiBin Sheng
- Department of Spine Surgery, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, China
| |
Collapse
|
27
|
DiNicolantonio JJ, McCarty MF, O'Keefe JH. Coenzyme Q10 deficiency can be expected to compromise Sirt1 activity. Open Heart 2022; 9:e001927. [PMID: 35296520 PMCID: PMC8928362 DOI: 10.1136/openhrt-2021-001927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
For reasons that remain unclear, endogenous synthesis and tissue levels of coenzyme Q10 (CoQ10) tend to decline with increasing age in at least some tissues. When CoQ10 levels are sufficiently low, this compromises the efficiency of the mitochondrial electron transport chain, such that production of superoxide by site 2 increases and the rate of adenosine triphosphate production declines. Moreover, CoQ10 deficiency can be expected to decrease activities of Sirt1 and Sirt3 deacetylases, believed to be key determinants of health span. Reduction of the cytoplasmic and mitochondrial NAD+/NADH ratio consequent to CoQ10 deficit can be expected to decrease the activity of these deacetylases by lessening availability of their obligate substrate NAD+ The increased oxidant production induced by CoQ10 deficiency can decrease the stability of Sirt1 protein by complementary mechanisms. And CoQ10 deficiency has also been found to lower mRNA expression of Sirt1. An analysis of the roles of Sirt1/Sirt3 in modulation of cellular function helps to rationalise clinical benefits of CoQ10 supplementation reported in heart failure, hypertension, non-alcoholic fatty liver disease, metabolic syndrome and periodontal disease. Hence, correction of CoQ10 deficiency joins a growing list of measures that have potential for amplifying health protective Sirt1/Sirt3 activities.
Collapse
Affiliation(s)
- James J DiNicolantonio
- Department of Preventive Cardiology, Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | | | - James H O'Keefe
- Saint Luke's Mid America Heart Institute, University of Missouri-Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
28
|
Tang L, Jiang J, Song G, Wang Y, Zhuang Z, Tan Y, Xia Y, Huang X, Feng X. Design, synthesis, and biological evaluation of novel urolithins derivatives as potential phosphodiesterase II inhibitors. Sci Rep 2021; 11:23792. [PMID: 34893678 PMCID: PMC8664850 DOI: 10.1038/s41598-021-03194-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/16/2021] [Indexed: 11/09/2022] Open
Abstract
A series of urolithins derivatives were designed and synthesized, and their structures have been confirmed by 1H NMR, 13C NMR, and HR-MS. The inhibitory activity of these derivatives on phosphodiesterase II (PDE2) was thoroughly studied with 3-hydroxy-8-methyl-6H-benzo[C]chromen-6-one and 3-hydroxy-7,8,9,10-tetrahydro-6H-benzo[C] chromen-6-one as the lead compounds. The biological activity test showed that compound 2e had the best inhibitory activity on PDE2 with an IC50 of 33.95 μM. This study provides a foundation for further structural modification and transformation of urolithins to obtain PDE2 inhibitor small molecules with better inhibitory activity.
Collapse
Affiliation(s)
- Long Tang
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing, 210042, China.,School of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China.,School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China
| | - Jianchun Jiang
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing, 210042, China.,School of Chemical Engineering, Nanjing Forestry University, Nanjing, 210037, China
| | - Guoqiang Song
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China
| | - Yajing Wang
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China
| | - Ziheng Zhuang
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China
| | - Ying Tan
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China
| | - Yan Xia
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China
| | - Xianfeng Huang
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China
| | - Xiaoqing Feng
- School of Pharmaceutical Engineering & Life Science, Changzhou University, Changzhou, 213164, China.
| |
Collapse
|